Liquid Biopsy for Lung Cancer: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , ,

Lung cancer is the deadliest cancer worldwide. Tissue biopsy is employed for the diagnosis and molecular stratification of lung cancer. Liquid biopsy is a minimally invasive approach to determine biomarkers from body fluids, such as blood, urine, sputum, and saliva. Tumor cells release cfDNA, ctDNA, exosomes, miRNAs, circRNAs, CTCs, and DNA methylated fragments, among others, which can be successfully used as biomarkers for diagnosis, prognosis, and prediction of treatment response.

  • liquid biopsy
  • lung cancer
  • biomarkers
  • precision medicine

1. Introduction

Lung cancer has the highest incidence rate for cancer type and the second-highest mortality rate in the world [1]. Liquid biopsy is a minimally invasive approach for sample procurement, mostly body fluids, and it is used to detect molecular alterations, tumor cells, and metabolites. Furthermore, liquid biopsy, the emerging approach attracting substantial attention, has currently been employed for clinical management, specifically for guiding treatment and monitoring disease. In addition, to being a minimally invasive approach, the liquid biopsy also enables the serial collection of samples allowing early detection of residual disease and relapses and resistance to treatment [2][3][4][5][6]. For non-small-cell lung cancer (NSCLC), this approach is particularly valuable for patients who are not eligible for the conventional tissue biopsy, mainly due to patients’ individual conditions and the tumor location, as previously discussed. As a result, liquid biopsy has been employed for disease and treatment monitoring and for creating a targeted treatment for NSCLC patients within the field of precision medicine [3][7].

2. NSCLC Biomarkers for Detection in Liquid Biopsy Samples

2.1. Circulating-Tumor DNA (ctDNA) and Cell-Free DNA (cfDNA)

Fragments of cell-free DNA (cfDNA) are freely available throughout the blood. CfDNA is released into the bloodstream due to natural body mechanisms, such as apoptosis, necrosis, and active secretion [5][8]. CfDNA can be found in both healthy subjects and cancer patients, although cfDNA levels tend to be higher in cancer patients.
Circulating tumor DNA (CtDNA), different from cfDNA, can harbor somatic mutations reflecting the tumor dynamics [9][10]. Tissue biopsy is a single “snapshot” of the tumor and, therefore, can mirror a unique subclone or a few subclones, while ctDNA can be more representative of the tumor’s entire tissue composition. Moreover, it is currently possible to trace the subclone’s origin of relapses and metastases through the phylogenetic profile of ctDNA. Tracing the clonal origin of the tumor can provide a collage of a tumor’s evolution, opening possibilities for the translation of such information into clinical practice [11].
Using liquid biopsy to detect ctDNA is extremely effective, as it is possible to collect a sample at any time during the disease and therapy course, assessing the progression of the disease in real-time [9]. However, liquid biopsy can become challenging when the concentration of ctDNA is measured to be extremely low (<1%) compared to the concentration of cfDNA in a patient’s bloodstream. Thence, highly sensitive and specific techniques are required for detecting mutations to track, detect, and monitor genomic alterations as cancer progresses [10][12][13]. The current standard methods for detecting somatic mutations, such as RT-PCR, Sanger sequencing, and next-generation sequencing (NGS), may not be sensitive enough for the detection of mutational ctDNA, specifically mutations presented with a low variant allele frequency (VAF) [9].
The main advantage of analyzing ctDNAs is the high specificity of the molecule, as it is proven that any molecular alterations present in these molecules are identical to those in the tumor tissue. In addition, advanced-stage cancer patients usually present with higher levels of ctDNA, which allows for easy monitoring of the disease’s course, tumor heterogeneity and dynamics, tumor evolution, and any tumor-acquired resistance to targeted treatments. Recently, ctDNA was associated with shorter survival, and actionable alterations in ctDNA were not detectable in time-matched tissue [14]. In addition, the turnaround time was decreased for the release of a biopsy report to guide therapeutic decisions that can be made earlier and safer [14].
Currently, highly sensitive PCR-based techniques, such as droplet digital PCR (ddPCR) and BEAMing, are considered sensitive enough for detecting mutations at low frequencies and have emerged as potential tools not only for advanced cancers but also for early detection [12]. NGS is also an extremely sensitive technique for detecting somatic mutations and identifying mutational frequencies as low as 0.02% VAF; however, the NGS technique produces an error rate of about 0.5–2.0% [15].
Therefore, the detection of rare variants by NGS remains challenging due to its limit of detection (LoD) and errors incorporated during sequencing. Furthermore, random errors can be incorporated into the DNA molecules during the library preparation or the sequencing processes, which could be mistaken for true variants. Strategies using molecular barcoding principles, such as hybrid capture and amplicon-based NGS, have also been used to detect low-frequency mutations in both liquid and conventional biopsy samples to decrease false negative results [9][12]. Each molecule in the sequencing library is marked with a small sequence of random nucleotides (8–16 N), which can be called a Unique Molecular Identifier (UMI), Tag Sequencing, or molecular barcodes and to decrease the chances of detecting false variants, the sequenced reads are grouped according to these UMI. As a result, sequencing artifacts can then be detected as they are not present in all reads with the same UMI, which increases the reliability of naming the true variants. Therefore, molecular barcoding technology allows for the correction of sequencing errors [16][17].
Due to the advances in sensitive technologies, ctDNA and mutational analysis are now possible for NSCLC patients. In addition, the detection rate of ctDNA can be higher than 80% in the plasma from NSCLC patients, suggesting that ctDNA analysis is an adequate alternative when sampling tissue biopsy is not an option [18]. EGFR, KRAS, ERBB2, and BRAF mutations, gene rearrangements (EML4—ALK, ROS1, NTRK1/2, and RET), exon skipping alterations, and gene amplifications (MET) are routinely evaluated in the management of care for NSCLC patients [9]. All these molecular alterations have been adapted into the clinical practice for guiding and monitoring patients’ treatment and disease state [12]. Currently, broader NGS panels have been employed in clinical practices, such as MSK-IMPACT (tissue) and MSK-ACCESS (plasma) [14]. A quarter of the patients presented alterations in ctDNA not detected in tissues [14], which suggests that plasma samples may present a higher specificity then previously reported, supporting the use of NGS as a sensitive, specific tool for plasma samples analysis [19].
Unfortunately, not all of the actionable alterations mentioned above have been translated for the clinical management of NSCLC patients through liquid biopsy sampling.
The FDA recently approved two IVD (in vitro diagnosis) tests (Cobas EGFR Mutation Test v2, Roche, and Idylla TM ctEGFR Mutation Assay) for NSCLC patients, which employs plasma samples for the detection of EGFR resistance mutation p.(Tyr790Met), exon 19 deletions and p/L858R mutations [20]. Although the IVD test shows a lower sensitivity when compared with other methods of variant detection (e.g., ddPCR and NGS), its approval was a great addition to the present tools used in guiding therapeutic decisions and monitoring treatment results for NSCLC patients [21].

2.2. cfDNA Methylation Biomarkers

Genetic mutations and epigenetic modifications, such as DNA methylation, have been detected in cfDNA and ctDNA. It has been considered a promising approach for translation into clinical applications to be used for diagnosis, prognosis, and predictive purposes.
Methylation is an incorporation of a methyl group (CH3) into a Cytosine in regions enriched with CG bases, also known as CpG islands [22]. Methylation occurs in CpG islands when found at the promoter regions of several genes, and it often results in gene silencing commonly found in tumor suppressor genes. On the other hand, the transcriptional activation of genes with the methylation present in the gene body is associated with various cancer types, including lung cancer [23][24]. Methylome analysis has yielded highly successful results on tumor tissues, especially when the analysis is focused on molecular subtyping and biomarkers discovery of several tumor types, including lung cancer [25][26]. In addition to plasma and serum, other body fluids deserve attention, such as sputum and bronchoalveolar lavage fluids, specifically due to their proximity to the tumor’s location [23].
Analysis of methylation-based biomarkers in cfDNA can also be used for diagnostic purposes for the management of lung cancer patients. Plasma cfDNA showed significant differences in DNA methylation level for early-stage NSCLC patients, including stage IA patients, indicating this type of biomarker could be a valuable tool for screening and early detection of NSCLC combined with imaging tests to improve the detection of early-stage pulmonary nodules [22][27][28][29][30]. Multi-cancer early detection (MCED) test may be a promising approach for cancer screening and early detection. MCED is a targeted methylation-based assay for complementary use in screening programs [31][32][33]. However, results for stage I lung cancer have not shown to be sensitive enough for being employed in a screening setting [32]. A recent study screened asymptomatic subjects from the NHS-Galleri trial (ISRCTN91431511) but results about the clinical utility of the MCED test for lung cancer still need to be addressed [33].
The combined methylation analysis of the CDO1 and HOXA9 was associated with unfavorable outcomes, while the combination of PTGDR and AJAP1 methylation was associated with favorable outcomes. A prognostic risk based on these methylation-based biomarkers may be useful to refine risk stratification [25][27]. In addition to prognostication, methylation-based biomarkers can also be useful as predictive biomarkers. Deregulation of methylation in cfDNA was also associated with EGFR-TKI resistance in early-stage NSCLC patients [34][35].

2.3. Circulating Tumor Cells (CTCs)

Circulating tumor cells (CTCs) are cells derived from primary tumors that were dissociated from tumor mass by either mechanical motion, the loss of adhesion molecules on the surface of cells entering the circulatory system, or a combination of both. CTCs are commonly detected in lower concentrations in the peripheral blood compared to other types of analytes (e.g., ctDNA), and it may be a challenge requiring the sampling of CTC-rich peripheral blood [10][13][36]. Although studies have shown that aggressive tumors can release thousands of these cells into the bloodstream every day, and it may be associated with the mechanism of distant metastasis, such a result would require completing a complex process [13][36]. Once these cells enter the bloodstream, they are capable of planting metastatic sites through active trans-endothelial migration while remaining inactive; however, no studies have yet been able to show how the biological process of transition from dormancy to active growth happens [10]. As a result, the mechanisms through which cancer spreads from one organ to another using CTCs are of great interest to the scientific community [13].
Furthermore, the methods of isolating CTCs include identifying these cells based on the presence of specific markers, such as the epithelial cell adhesion molecule (Ep-CAM), the cytokeratin of epithelial CTCs, and the N-Cadherin or vimentin of mesenchymal CTCs [37][38]. The FDA has confirmed an IVD method, based on anti-EpCAM ferromagnetic microbeads, called CellSearch CTC kit® (Veridex LLC, Raritan, NJ, USA) for prognostic assessment. This IVD method detects anti-EpCAM ferromagnetic microbeads on circulating tumor cells (CTC) in peripheral blood samples [39]. As previously mentioned, the characterization of CTCs is difficult since the detection method requires high sensitivity to detect these molecules’ low concentrations in extracorporeal fluids.
The presence of CTCs is considered a prognostic biomarker since it can help predict disease progression in cancer patients, including the progression of NSCLC [13]. One study showed that patients with metastatic lung cancer and a state of progressive disease presented a higher expression of the PIK3CA, AKT2, TWIST, and ALDH1 genes in CTCs compared to patients with non-metastatic disease. Thus, it is possible to correlate the presence of CTCs with the diagnostic scope of disease burden, including possible metastasis and disease progression [40]. The presence of CTCs was found to be independent of the tumor stage at diagnosis (49% stage I, 48% stage II, 48% stage III, and 52% stage IV patients) and histology (47% adenocarcinoma and 40% squamous cell carcinoma) when using a size-based filtration method for CTC detection [41]. Moreover, of the cells with the highest glucose uptake, hypermetabolic CTCs were isolated to analyze EGFR and KRAS mutations using ddPCR. The comparison between the primary tumor’s EGFR and KRAS mutations and that of CTCs showed a match in 70% of the cases [42].

2.4. Extracellular Vesicles

Extracellular vesicles (EVs) can be divided into microvesicles, vesicles, and exosomes. Exosomes are released by various methods and are detectable in several cell types and bodily fluids, including cancer cells. Moreover, exosomes are released during the process of exocytosis following the fusion of multivesicular bodies (MVBs) and cell membranes, and they can also be detected in body fluids such as blood (plasma and serum), urine, pleural effusions, saliva, cerebrospinal fluid, and semen [6][13]. In addition, exosomes have been shown to mediate intercellular communication between the tumor and the stroma, resulting in a rich source of molecular information that enables the location of origin cells for these exosomes [43][44].
Furthermore, ultracentrifugation or differential centrifugation (DC) and size-exclusion chromatography, based on size selection and chemical isolation during polymeric-based precipitation (PBP), are all commonly used for EV isolation [45][46][47].
Tumor cells can release more exosomes than non-tumor cells, making exosomes a potential biomarker in liquid biopsies for various types of tumors [6]. In lung cancer, exosomal RNA, DNA, and proteins can be used to detect molecular alterations, including actionable mutations [6][48]. Qu et al. (2019) identified exosomes harboring EGFR mutation, showcasing exosomes as a valuable component in the analysis’s tumor progression, pre-metastatic niche formation, and resistance to treatment [49]. The comparison between exosome-derived and tumor-derived mutations showed a sensitivity of 100% in detecting EGFR mutations and a specificity greater than 96% [49].
Furthermore, exosomal miRNAs (exo-miRNA) have also been generating interest from the scientific community as a potential biomarker in several types of cancer, including lung cancer. Exo-miRNA is protected from RNAse degradation in the bloodstream due to the lipid bilayer membrane, which results in their stability in body fluids and easily detectable form in body fluids [6]. As a result, exo-miRNA load was reported as a prognostic biomarker [50]. Additionally, exo-miRNAs miR-564 and miR-659 switched sensitive to resistant cells to gefitinib [51], which proved to be a predictive phenotype for exo-miRNA. On the other hand, the exo-miRNA miR-302-b is associated with the suppression of lung cancer cell proliferation and migration via the TGFβRII/ERK pathways, which has established the miR-302-b as a potential therapeutic target for lung cancer patients [52]. Thus, exosomal miRNAs may be employed in the process and measurements of diagnosis, prognosis, and monitoring of lung cancer patients, which reveals the need for further studies of exosomes and their clinical application in the context of precision medicine.

2.5. MicroRNA/CircRNA

MicroRNAs (miRNAs) are small RNAs (19–24 nucleotides) responsible for regulating gene expression. However, most miRNAs have unknown biological functions [53][54]. MiRNAs differ from mRNAs due to their stability, small size, and regulatory control in gene expression, rendering them a new generation of biomarkers [55]. Circulating-free miRNAs (cfmiRNAs) have already been reported in body fluids from cancer patients, including plasma, serum, urine, and saliva [56][57][58].
The detection of miRNAs in liquid biopsy samples was reported to distinguish NSCLC patients from healthy subjects paving the way for cfmiRNAs as promising early detection biomarkers [59]. Reis and collaborators (2020) recently reported differential expression of a set of miRNAs in plasma samples from early-stage lung cancer patients [60]. Similarly, plasma samples from early-stage NSCLC patients paired with non-cancer patients were screened for 754 circulating miRNAs, and a highly accurate 24-miRNAs panel was proposed for early detection of lung cancer in addition to the known risk factors [59]. In addition, dysregulation of miRNA expression can also be associated with exposure factors.
For example, the downregulation of let-7i-3p and miR-154-5p was found in serum from smokers, both non-cancer subjects and lung cancer patients. These miRNAs are involved in lung cancer development and progression, rendering these circulating miRNAs a potential role as a diagnostic and prognostic biomarker in lung cancer for tobacco-exposed patients [61][62]. Moreover, high expression of miR-34 and miR34-c in plasma samples from surgically resected NSCLC patients was associated with increased survival [62].
Recently, some publications have explored the potential of circular RNA (circRNA) as a biomarker in liquid biopsy [63]. This is because circRNA are resistant to RNases and other exonucleases due to the lack of final 5’ or 3’ regions [64][65], having a longer half-life than linear RNA [66]. Moreover, its specific tissue expression and stage of development increase this interest as a potential biomarker [62][67][68][69]. Studies show that circRNA expression varies in different tumor types, including lung cancer [68][70][71][72]. In adenocarcinoma patients, circRNA can be over-expressed, such as circ_0013958, which was upregulated [71], and circFARSA, a circRNA derived from exon 5–7 of the FARSA gene, in the plasma of patients with NSCLC compared to controls without cancer [73][74].
In addition, studies with circRNAs reported that they could detect specific signature profiles in tissue samples, capable of distinguishing histological groups in addition to NCLSC patients from healthy ones [69]. However, studies are being carried out to analyze the profile of circRNAs signatures in liquid biopsy samples, such as plasma, exosomes, and lymphocytes, to implement these molecules as diagnostic and predictive biomarkers.
Altogether, the detection of miRNAs/circRNAs in minimally invasive samples has emerged as a promising approach for the early detection of NSCLC to be incorporated in lung cancer screening programs and for prognostic purposes to be incorporated in the clinical management of these patients.

2.6. Metabolites and Proteins

In pathological conditions, including cancer, circulating metabolites can be detected in body fluids [75]. For detecting metabolites in biological fluids, Gas chromatography-mass spectrometry (GC-MS), nuclear magnetic resonance (NMR), and high-performance liquid chromatography UV detector (LC-UV) along with software (Metaboanalyst, Metabolite Set Enrichment Analysis [MSEA], Metlin, BioStatFlow and Human metabolome database [HMDB]) have been employed for detecting, processing, and analyzing metabolic data [76].
Lung cancer patients show changes in metabolic pathways compared to non-cancer patients; starch and sucrose metabolism, galactose metabolism, fructose, mannose degradation, purine metabolism, and tryptophan metabolism are among the unbalanced metabolic pathways [77]. In addition, some amino acids such as valine, leucine, and isoleucine are related to stress and energy production, regulating many signaling pathways, such as protein synthesis, lipid synthesis, cell growth, and autophagy, and can be found at higher levels in lung cancer patients compared with non-cancer patients [78]. However, these previously reported data must be clinically tested and reproduced in other series.
Although metabolomics has been a promising approach for managing and monitoring cancer patients, some limitations have still been experienced, such as identifying unknown compounds, detecting cancer-specific metabolites, and standardization of cutoffs [76].
In addition to metabolites, serum proteins have currently been used as cancer biomarkers, such as carcinoembryonic antigen (CEA), cytokeratin 19fragment (CYFRA 21-1), cancer antigen 125 (CA 125), neuron-specific enolase (NSE) and squamous cell carcinoma antigen (SCCA) [79][80]. Immunoassay (e.g., ELISA) and mass spectrometry can detect these serum proteins. In the last few years, considerable efforts have been made to find serum protein biomarkers for the early detection of lung cancer that will potentially be employed in screening programs soon.
Acute-phase reactant proteins (APRPs) are produced in response to inflammation caused by cancer and can also be used as potential biomarkers for the diagnosis of different types of cancer [81]. Lung cancer patients presented higher serum haptoglobin β (HP- β) chain levels than healthy controls. However, patients with other respiratory diseases also presented increased levels of Hp-β chain; thus, medical history, and radiological images should also be considered [82]. Lung cancer patients also presented higher serum amyloid A (SAA), another APRP, compared with healthy controls [83]. With higher levels of SAA1 and SAA2, lung adenocarcinoma patients also presented decreased serum levels of Apo A-1—a protein responsible for removing endogenous cholesterol from inflammatory sites. Thus, SAA1, SAA2, and Apo A-1 could also be considered potential biomarkers for the early detection of lung cancer [84].
In addition, to early detection applications, some proteins involved with metastasis and tumor progression are promising prognostic biomarkers. For example, plasma and pleural effusions from NSCLC patients with high levels of the S100A6 protein—a member of the S100 family with pro-apoptotic function—presented longer survival time compared with S100A6-negative cases [85][86]. On the other hand, NSCLC patients show high serum Cytokeratins (CKs) levels, such as CK 8, 18, and 19, which were associated with unfavorable prognoses [86][87]. In addition, the combined analysis of regulators of actin—calmodulin, thymosin β4, cofilin-1, and thymosin β10—was suitable for predicting patients’ outcomes [88].
The CancerSEEK reported forty-one potential protein biomarkers detectable at least in one out of the eight tumor types. The authors selected the best eight biomarkers for composing the final bead-based immunoassay test (CA-125, CA19-9, CEA, HGF, Myeloperoxidase, OPN, Prolactin, TIMP-1), which were highly effective in distinguishing cancer patients from healthy controls [89]. The positivity for the CancerSEEK test was about 70% for all cancer types. Combining protein biomarkers with cfDNA mutations increased sensitivity without significantly decreasing specificity [89].
Although proteomic approaches have been promising tools for precision medicine in the lung cancer field, there are some limitations regarding using proteins as biomarkers. Many of these proteins are associated with different tumor types. They present poor sensitivity, are not organ-specific, and can also be detected in non-malignant diseases [79][80][90]. Thus, clinical history and radiological exams should always be considered. Moreover, some proteins are presented with low serum levels, precluding their employment for early lung cancer diagnosis [90][91]. Finally, the analysis of these biomarkers can be influenced by the contamination of intracellular proteins caused by cell lysis during sample procurement and processing. Thus, pre-analytical steps are pivotal [85].
Scalable proteomics has also emerged as a tool for the identification of high-risk subjects for lung cancer. The most recent example is Olink Proteomics, which was created to detect circulating proteins (https://olink.com/, accessed on 16 November 2022). A 36-protein multiplexed assay was developed for risk assessment of lung cancer development, and this set of proteins includes growth factors, tumor necrosis factor receptors, and chemokines and cytokines [92]. Scalable proteomic approaches may not be affordable for low-middle-income countries, but when feasible, they should still be considered as a complementary tool along with LDCT in lung cancer screening programs. However, more studies should be conducted to prove these proteomic approaches are cost-effective for lung cancer screening, especially considering limited resources and minorities.

2.7. Autoantibodies against Tumor-Associated Antigens

Autoantibodies against tumor-associated antigens (TAAs) are circulating antibodies to autologous cellular antigens [93][94][95]. Tumor tissues can release cellular proteins, leading to the activation of the immune system and the production of autoantibodies [96][97]. Therefore, cancer patients produce autoantibodies against aberrant or overexpressed proteins produced by these cancer cells [98]. TAAs are stable in the serum and have been studied in several types of tumors, including lung cancer. TAAs can be detected through the immunoenzymatic assay (ELISA) [96][99][100]. Moreover, they can also provide information for early detection and disease monitoring [99][100][101][102][103][104].
In the 90s, Lubin et al. (1995) analyzed the presence of p53 antibodies in the serum of patients with lung cancer [105]. These antibodies were 30% higher in cancer patients than in non-cancer patients, associated with TP53 mutations [105]. TAAs panels have also been developed, combining many different TAAs to improve test sensitivity [93][99][100][106][107]. The panels developed include TAAs for p53, NY-ESO-1, CAGE, GBU4–5, Annexin 1, SOX2, c-Myc, MDM2, NPM1, p16, cyclin B1, among others [99][100][108]. Although the levels of most biomarkers increase proportionally to tumor burden, TAAs levels do not differ among different stages of lung cancer, possibly due to the humoral immune response—present from the beginning of tumorigenesis [99][100][108]. This feature renders TAAs a potential tool for early detection [99][100][109][110]. However, studies including proper sample size and validation set are required for greater reliability of TAAs in the routine lung cancer setting.

This entry is adapted from the peer-reviewed paper 10.3390/ijms24032505

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34.
  2. Lim, M.; Kim, C.-J.; Sunkara, V.; Kim, M.-H.; Cho, Y.-K. Liquid Biopsy in Lung Cancer: Clinical Applications of Circulating Biomarkers (CTCs and ctDNA). Micromachines 2018, 9, 100.
  3. Lu, J.; Han, B. Liquid Biopsy Promotes Non-Small Cell Lung Cancer Precision Therapy. Technol. Cancer Res. Treat. 2018, 17, 1533033818801809.
  4. Hoseok, I.; Cho, J.-Y. Lung cancer biomarkers. Adv. Clin. Chem. 2015, 72, 107–170.
  5. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238.
  6. Bracht, J.W.P.; Mayo-De-Las-Casas, C.; Berenguer, J.; Karachaliou, N.; Rosell, R. The Present and Future of Liquid Biopsies in Non-Small Cell Lung Cancer: Combining Four Biosources for Diagnosis, Prognosis, Prediction, and Disease Monitoring. Curr. Oncol. Rep. 2018, 20, 70.
  7. Trombetta, D.; Sparaneo, A.; Fabrizio, F.P.; Muscarella, L.A. Liquid biopsy and NSCLC. Lung Cancer Manag. 2016, 5, 91–104.
  8. Wang, Y.; Springer, S.; Zhang, M.; McMahon, K.W.; Kinde, I.; Dobbyn, L.; Ptak, J.; Brem, H.; Chaichana, K.; Gallia, G.L.; et al. Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord. Proc. Natl. Acad. Sci. USA 2015, 112, 9704–9709.
  9. Diaz, L.A., Jr.; Bardelli, A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2014, 32, 579.
  10. Davis, A.A.; Cristofanilli, M. Detection of Predictive Biomarkers Using Liquid Biopsies. In Predictive Biomarkers in Oncology; Springer: Berlin/Heidelberg, Germany, 2019; pp. 107–117.
  11. Abbosh, C.; Birkbak, N.J.; Wilson, G.A.; Jamal-Hanjani, M.; Constantin, T.; Salari, R.; Le Quesne, J.; Moore, D.A.; Veeriah, S.; Rosenthal, R.; et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017, 545, 446–451.
  12. Gale, D.; Lawson, A.R.; Howarth, K.; Madi, M.; Durham, B.; Smalley, S.; Calaway, J.; Blais, S.; Jones, G.; Clark, J.; et al. Development of a highly sensitive liquid biopsy platform to detect clinically-relevant cancer mutations at low allele fractions in cell-free DNA. PLoS ONE 2018, 13, e0194630.
  13. Revelo, A.E.; Martin, A.; Velasquez, R.; Kulandaisamy, P.C.; Bustamante, J.; Keshishyan, S.; Otterson, G. Liquid biopsy for lung cancers: An update on recent developments. Ann. Transl. Med. 2019, 7, 349.
  14. Jee, J.; Lebow, E.S.; Murciano-Goroff, Y.R.; Jayakumaran, G.; Shen, R.; Brannon, A.R.; Benayed, R.; Namakydoust, A.; Offin, M.; Paik, P.K.; et al. Overall Survival with Circulating Tumor DNA-Guided Therapy in Advanced Non-Small Cell Lung Cancer; Wolters Kluwer Health: Philadelphia, PA, USA, 2021.
  15. Shendure, J.; Ji, H. Next-generation DNA sequencing. Nat. Biotechnol. 2008, 26, 1135–1145.
  16. Wong, W.H.; Tong, R.S.; Young, A.L.; Druley, T.E. Rare Event Detection Using Error-corrected DNA and RNA Sequencing. J. Vis. Exp. 2018, e57509.
  17. Teder, H.; Koel, M.; Paluoja, P.; Jatsenko, T.; Rekker, K.; Laisk-Podar, T.; Kukuškina, V.; Velthut-Meikas, A.; Fjodorova, O.; Peters, M.; et al. TAC-seq: Targeted DNA and RNA sequencing for precise biomarker molecule counting. NPJ Genom. Med. 2018, 3, 34.
  18. Villaflor, V.; Won, B.; Nagy, R.; Banks, K.; Lanman, R.B.; Talasaz, A.; Salgia, R. Biopsy-free circulating tumor DNA assay identifies actionable mutations in lung cancer. Oncotarget 2016, 7, 66880.
  19. Sabari, J.K.; Offin, M.; Stephens, D.; Ni, A.; Lee, A.; Pavlakis, N.; Clarke, S.; Diakos, C.I.; Datta, S.; Tandon, N.; et al. A Prospective Study of Circulating Tumor DNA to Guide Matched Targeted Therapy in Lung Cancers. Gynecol. Oncol. 2019, 111, 575–583.
  20. FuFaD, A. Aprovação Pré-Comercialização P150044, Teste de Mutação Cobas EGFR V2. 2017 FD. 2017. Available online: https://www.accessdata.fda.gov/cdrh_docs/pdf12/p120019s007c.pdf (accessed on 1 September 2022).
  21. Torres, S.; González, Á.; Tomás, A.J.C.; Fariñas, S.C.; Ferrero, M.; Mirda, D.; Sirera, R.; Jantus-Lewintre, E.; Camps, C. A profile on cobas® EGFR Mutation Test v2 as companion diagnostic for first-line treatment of patients with non-small cell lung cancer. Expert Rev. Mol. Diagn. 2020, 20, 575–582.
  22. Vrba, L.; Oshiro, M.M.; Kim, S.S.; Garland, L.L.; Placencia, C.; Mahadevan, D.; Nelson, M.A.; Futscher, B.W. DNA methylation biomarkers discovered in silico detect cancer in liquid biopsies from non-small cell lung cancer patients. Epigenetics 2020, 15, 419–430.
  23. Li, L.; Fu, K.; Zhou, W.; Snyder, M. Applying circulating tumor DNA methylation in the diagnosis of lung cancer. Precis. Clin. Med. 2019, 2, 45–56.
  24. Duruisseaux, M.; Esteller, M. (Eds.) Lung cancer epigenetics: From knowledge to applications. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2018.
  25. Bjaanaes, M.M.; Fleischer, T.; Halvorsen, A.R.; Daunay, A.; Busato, F.; Solberg, S.; Jørgensen, L.; Kure, E.; Edvardsen, H.; Børresen-Dale, A.-L.; et al. Genome-wide DNA methylation analyses in lung adenocarcinomas: Association with EGFR, KRAS and TP53 mutation status, gene expression and prognosis. Mol. Oncol. 2016, 10, 330–343.
  26. Wang, J.; Duan, Y.; Meng, Q.-H.; Gong, R.; Guo, C.; Zhao, Y.; Zhang, Y. Integrated analysis of DNA methylation profiling and gene expression profiling identifies novel markers in lung cancer in Xuanwei, China. PLoS ONE 2018, 13, e0203155.
  27. Ooki, A.; Maleki, Z.; Tsay, J.-C.J.; Goparaju, C.; Brait, M.; Turaga, N.; Nam, H.-S.; Rom, W.N.; Pass, H.I.; Sidransky, D.; et al. A Panel of Novel Detection and Prognostic Methylated DNA Markers in Primary Non–Small Cell Lung Cancer and Serum DNA. Clin. Cancer Res. 2017, 23, 7141–7152.
  28. Yang, Z.; Qi, W.; Sun, L.; Zhou, H.; Zhou, B.; Hu, Y. DNA methylation analysis of selected genes for the detection of early-stage lung cancer using circulating cell-free DNA. Adv. Clin. Exp. Med. Off. Organ Wroc. Med. Univ. 2019, 28, 355–360.
  29. Pérez-Ramírez, C.; Cañadas-Garre, M.; Robles, A.I.; Molina, M.; Faus-Dáder, M.J.; Calleja-Hernández, M. Liquid biopsy in early stage lung cancer. Transl. Lung Cancer Res. 2016, 5, 517.
  30. Hulbert, A.; Jusue-Torres, I.; Stark, A.; Chen, C.; Rodgers, K.; Lee, B.; Griffin, C.; Yang, A.; Huang, P.; Wrangle, J.; et al. Early Detection of Lung Cancer Using DNA Promoter Hypermethylation in Plasma and SputumEpigenetic Lung Cancer Screening. Clin. Cancer Res. 2017, 23, 1998–2005.
  31. Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Swanton, C.; Seiden, M.V.; CCGA Consortium. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann. Oncol. 2020, 31, 745–759.
  32. Klein, E.; Richards, D.; Cohn, A.; Tummala, M.; Lapham, R.; Cosgrove, D.; Chung, G.; Clement, J.; Gao, J.; Hunkapiller, N.; et al. Clinical validation of a targeted methylation-based multi-cancer early detection test using an independent validation set. Ann. Oncol. 2021, 32, 1167–1177.
  33. Neal, R.D.; Johnson, P.; Clarke, C.A.; Hamilton, S.A.; Zhang, N.; Kumar, H.; Swanton, C.; Sasieni, P. Cell-Free DNA–Based Multi-Cancer Early Detection Test in an Asymptomatic Screening Population (NHS-Galleri): Design of a Pragmatic, Prospective Randomised Controlled Trial. Cancers 2022, 14, 4818.
  34. Poggiana, C.; Rossi, E.; Zamarchi, R. Possible role of circulating tumor cells in early detection of lung cancer. J. Thorac. Dis. 2020, 12, 3821.
  35. Santarpia, M.; Liguori, A.; Karachaliou, N.; Gonzalez-Cao, M.; Daffinà, M.G.; D’Aveni, A.; Marabello, G.; Altavilla, G.; Rosell, R. Osimertinib in the treatment of non-small-cell lung cancer: Design, development and place in therapy. Lung Cancer Targets Ther. 2017, 8, 109.
  36. Shaffer, C. Circulating Tumor Cells and the Liquid Biopsy: Processed appropriately, liquid biopsy samples and circulating biomarkers may aid cancer diagnosis and predict treatment outcomes. Genet. Eng. Biotechnol. News 2019, 39, 42–44.
  37. He, Y.; Shi, J.; Schmidt, B.; Liu, Q.; Shi, G.; Xu, X.; Liu, C.; Gao, Z.; Guo, T.; Shan, B. Circulating Tumor Cells as a Biomarker to Assist Molecular Diagnosis for Early Stage Non-Small Cell Lung Cancer. Cancer Manag. Res. 2020, 12, 841.
  38. Correnti, M.; Raggi, C. Stem-like plasticity and heterogeneity of circulating tumor cells: Current status and prospect challenges in liver cancer. Oncotarget 2017, 8, 7094.
  39. Zhang, Y.; Wang, Y.; Du, Z.; Wu, M.; Zhang, G. Detection of micrometastases in lung cancer with magnetic nanoparticles and quantum dots. Int. J. Nanomed. 2012, 7, 2315.
  40. Hanssen, A.; Wagner, J.; Gorges, T.M.; Taenzer, A.; Uzunoglu, F.G.; Driemel, C.; Stoecklein, N.H.; Knoefel, W.T.; Angenendt, S.; Hauch, S.; et al. Characterization of different CTC subpopulations in non-small cell lung cancer. Sci. Rep. 2016, 6, 28010.
  41. Tu, Q.; Wu, X.; Le Rhun, E.; Blonski, M.; Wittwer, B.; Taillandier, L.; Bittencourt, M.D.C.; Faure, G.C. CellSearch® technology applied to the detection and quantification of tumor cells in CSF of patients with lung cancer leptomeningeal metastasis. Lung Cancer 2015, 90, 352–357.
  42. Hofman, V.; Long, E.; Ilie, M.; Bonnetaud, C.; Vignaud, J.M.; Fléjou, J.F.; Lantuejoul, S.; Piaton, E.; Mourad, N.; Butori, C.; et al. Morphological analysis of circulating tumour cells in patients undergoing surgery for non-small cell lung carcinoma using the isolation by size of epithelial tumour cell (ISET) method. Cytopathology 2012, 23, 30–38.
  43. Miyanaga, A.; Masuda, M.; Yamada, T. Biomarkers of Lung Cancer: Liquid Biopsy Comes of Age. In Biomarkers in Cancer Therapy; Springer: Berlin/Heidelberg, Germany, 2019; pp. 105–113.
  44. Turetta, M.; Bulfoni, M.; Brisotto, G.; Fasola, G.; Zanello, A.; Biscontin, E.; Mariuzzi, L.; Steffan, A.; Di Loreto, C.; Cesselli, D.; et al. Assessment of the Mutational Status of NSCLC Using Hypermetabolic Circulating Tumor Cells. Cancers 2018, 10, 270.
  45. Reclusa, P.; Taverna, S.; Pucci, M.; Durendez, E.; Calabuig, S.; Manca, P.; Serrano, M.J.; Sober, L.; Pauwels, P.; Russo, A.; et al. Exosomes as diagnostic and predictive biomarkers in lung cancer. J. Thorac. Dis. 2017, 9 (Suppl. S13), S1373–S1382.
  46. Vanni, I.; Alama, A.; Grossi, F.; Dal Bello, M.G.; Coco, S. Exosomes: A new horizon in lung cancer. Drug Discov. Today 2017, 22, 927–936.
  47. Momen-Heravi, F.; Balaj, L.; Alian, S.; Mantel, P.-Y.; Halleck, A.E.; Trachtenberg, A.J.; Soria, C.E.; Oquin, S.; Bonebreak, C.M.; Saracoglu, E.; et al. Current methods for the isolation of extracellular vesicles. Biol. Chem. 2013, 394, 1253–1262.
  48. Böing, A.N.; van der Pol, E.; Grootemaat, A.E.; Coumans, F.A.W.; Sturk, A.; Nieuwland, R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 2014, 3, 23430.
  49. Qu, X.; Li, Q.; Yang, J.; Zhao, H.; Wang, F.; Zhang, F.; Zhang, S.; Zhang, H.; Wang, R.; Wang, Q.; et al. Double-Stranded DNA in Exosomes of Malignant Pleural Effusions as a Novel DNA Source for EGFR Mutation Detection in Lung Adenocarcinoma. Front. Oncol. 2019, 9, 931.
  50. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 14, 531–548.
  51. Rodríguez, M.; Silva, J.; López-Alfonso, A.; López-Muñiz, M.B.; Peña, C.; Domínguez, G.; García, J.M.; López-Gónzalez, A.; Méndez, M.; Provencio, M.; et al. Different exosome cargo from plasma/bronchoalveolar lavage in non-small-cell lung cancer. Genes Chromosom. Cancer 2014, 53, 713–724.
  52. Rabinowits, G.; Gerçel-Taylor, C.; Day, J.M.; Taylor, D.D.; Kloecker, G.H. Exosomal MicroRNA: A Diagnostic Marker for Lung Cancer. Clin. Lung Cancer 2009, 10, 42–46.
  53. Tomasetti, M.; Lee, W.; Santarelli, L.; Neuzil, J. Exosome-derived microRNAs in cancer metabolism: Possible implications in cancer diagnostics and therapy. Exp. Mol. Med. 2017, 49, e285.
  54. Galka-Marciniak, P.; Urbanek-Trzeciak, M.O.; Nawrocka, P.M.; Dutkiewicz, A.; Giefing, M.; Lewandowska, M.A.; Kozlowski, P. Somatic Mutations in miRNA Genes in Lung Cancer—Potential Functional Consequences of Non-Coding Sequence Variants. Cancers 2019, 11, 793.
  55. Iqbal, M.A.; Arora, S.; Prakasam, G.; Calin, G.A.; Syed, M.A. MicroRNA in lung cancer: Role, mechanisms, pathways and therapeutic relevance. Mol. Asp. Med. 2019, 70, 3–20.
  56. Almeida, M.I.; Reis, R.M.; Calin, G.A. MicroRNA history: Discovery, recent applications, and next frontiers. Mutat. Res./Fundam. Mol. Mech. Mutagen. 2011, 717, 1–8.
  57. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518.
  58. Puerta-Gil, P.; García-Baquero, R.; Jia, A.Y.; Ocaña, S.; Alvarez-Múgica, M.; Alvarez-Ossorio, J.L.; Cordon-Cardo, C.; Cava, F.; Sánchez-Carbayo, M. miR-143, miR-222, and miR-452 Are Useful as Tumor Stratification and Noninvasive Diagnostic Biomarkers for Bladder Cancer. Am. J. Pathol. 2012, 180, 1808–1815.
  59. Zhi, F.; Cao, X.; Xie, X.; Wang, B.; Dong, W.; Gu, W.; Ling, Y.; Wang, R.; Yang, Y.; Liu, Y. Identification of Circulating MicroRNAs as Potential Biomarkers for Detecting Acute Myeloid Leukemia. PLoS ONE 2013, 8, e56718.
  60. Reis, P.P.; Drigo, S.A.; Carvalho, R.F.; Lapa, R.M.L.; Felix, T.F.; Patel, D.; Cheng, D.; Pintilie, M.; Liu, G.; Tsao, M.-S. Circulating miR-16-5p, miR-92a-3p, and miR-451a in Plasma from Lung Cancer Patients: Potential Application in Early Detection and a Regulatory Role in Tumorigenesis Pathways. Cancers 2020, 12, 2071.
  61. Wozniak, M.B.; Scelo, G.; Muller, D.C.; Mukeria, A.; Zaridze, D.; Brennan, P. Circulating MicroRNAs as Non-Invasive Biomarkers for Early Detection of Non-Small-Cell Lung Cancer. PLoS ONE 2015, 10, e0125026.
  62. Huang, J.; Wu, J.; Li, Y.; Li, X.; Yang, T.; Yang, Q.; Jiang, Y. Deregulation of Serum MicroRNA Expression Is Associated with Cigarette Smoking and Lung Cancer. BioMed Res. Int. 2014, 2014, 364316.
  63. Zhao, K.; Cheng, J.; Chen, B.; Liu, Q.; Xu, D.; Zhang, Y. Circulating microRNA-34 family low expression correlates with poor prognosis in patients with non-small cell lung cancer. J. Thorac. Dis. 2017, 9, 3735.
  64. Arnaiz, E.; Sole, C.; Manterola, L.; Iparraguirre, L.; Otaegui, D.; Lawrie, C.H. (Eds.) CircRNAs and cancer: Biomarkers and master regulators. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2019.
  65. Suzuki, H.; Zuo, Y.; Wang, J.; Zhang, M.Q.; Malhotra, A.; Mayeda, A. Characterization of RNase R-digested cellular RNA source that consists of lariat and circular RNAs from pre-mRNA splicing. Nucleic Acids Res. 2006, 34, e63.
  66. Vincent, H.A.; Deutscher, M.P. Substrate Recognition and Catalysis by the Exoribonuclease RNase R. J. Biol. Chem. 2006, 281, 29769–29775.
  67. Salzman, J.; Chen, R.E.; Olsen, M.N.; Wang, P.L.; Brown, P.O. Cell-type specific features of circular RNA expression. PLoS Genet. 2013, 9, e1003777.
  68. Jeck, W.R.; Sharpless, N.E. Detecting and characterizing circular RNAs. Nat. Biotechnol. 2014, 32, 453–461.
  69. De Fraipont, F.; Gazzeri, S.; Cho, W.C.; Eymin, B. Circular RNAs and RNA splice variants as biomarkers for prognosis and therapeutic response in the liquid biopsies of lung cancer patients. Front. Genet. 2019, 10, 390.
  70. Pedraz-Valdunciel, C.; Giannoukakos, S.; Potie, N.; Giménez-Capitán, A.; Huang, C.; Hackenberg, M.; Fernandez-Hilario, A.; Bracht, J.; Filipska, M.; Aldeguer, E.; et al. Digital multiplexed analysis of circular RNAs in FFPE and fresh non-small cell lung cancer specimens. Mol. Oncol. 2022, 16, 2367–2383.
  71. Chen, K.-Z.; Lou, F.; Yang, F.; Zhang, J.-B.; Ye, H.; Chen, W.; Guan, T.; Zhao, M.-Y.; Su, X.-X.; Shi, R.; et al. Circulating Tumor DNA Detection in Early-Stage Non-Small Cell Lung Cancer Patients by Targeted Sequencing. Sci. Rep. 2016, 6, 31985.
  72. Zhu, X.; Wang, X.; Wei, S.; Chen, Y.; Chen, Y.; Fan, X.; Han, S.; Wu, G. hsa_circ_0013958: A circular RNA and potential novel biomarker for lung adenocarcinoma. FEBS J. 2017, 284, 2170–2182.
  73. Zhang, X.; Zhou, H.; Jing, W.; Luo, P.; Qiu, S.; Liu, X.; Zhu, M.; Liang, C.; Yu, M.; Tu, J. The Circular RNA hsa_circ_0001445 Regulates the Proliferation and Migration of Hepatocellular Carcinoma and May Serve as a Diagnostic Biomarker. Dis. Markers 2018, 2018, 3073467.
  74. Hang, D.; Zhou, J.; Qin, N.; Zhou, W.; Ma, H.; Jin, G.; Hu, Z.; Dai, J.; Shen, H. A novel plasma circular RNA circFARSA is a potential biomarker for non-small cell lung cancer. Cancer Med. 2018, 7, 2783–2791.
  75. Seow, W.J.; Shu, X.-O.; Nicholson, J.; Holmes, E.; Walker, D.I.; Hu, W.; Cai, Q.; Gao, Y.-T.; Xiang, Y.-B.; Moore, S.C.; et al. Association of Untargeted Urinary Metabolomics and Lung Cancer Risk Among Never-Smoking Women in China. JAMA Netw. Open 2019, 2, e1911970.
  76. Yu, L.; Li, K.; Zhang, X. Next-generation metabolomics in lung cancer diagnosis, treatment and precision medicine: Mini review. Oncotarget 2017, 8, 115774.
  77. Sun, Q.; Zhao, W.; Wang, L.; Guo, F.; Song, D.; Zhang, Q.; Zhang, D.; Fan, Y.; Wang, J. Integration of metabolomic and transcriptomic profiles to identify biomarkers in serum of lung cancer. J. Cell. Biochem. 2019, 120, 11981–11989.
  78. Tang, Y.; Li, Z.; Lazar, L.; Fang, Z.; Tang, C.; Zhao, J. Metabolomics workflow for lung cancer: Discovery of biomarkers. Clin. Chim. Acta 2019, 495, 436–445.
  79. Kumar, A.; Misra, B.B. Challenges and Opportunities in Cancer Metabolomics. Proteomics 2019, 19, e1900042.
  80. Molina, R.; Filella, X.; Augé, J.; Fuentes, R.; Bover, I.; Rifa, J.; Moreno, V.; Canals, E.; Viñolas, N.; Marquez, A.; et al. Tumor Markers (CEA, CA 125, CYFRA 21-1, SCC and NSE) in Patients with Non-Small Cell Lung Cancer as an Aid in Histological Diagnosis and Prognosis. Tumor Biol. 2003, 24, 209–218.
  81. Barlési, F.; Gimenez, C.; Torre, J.-P.; Doddoli, C.; Mancini, J.; Greillier, L.; Roux, F.; Kleisbauer, J.-P. Prognostic value of combination of Cyfra 21-1, CEA and NSE in patients with advanced non-small cell lung cancer. Respir. Med. 2004, 98, 357–362.
  82. Pang, W.W.; Abdul-Rahman, P.S.; Wan-Ibrahim, W.I.; Hashim, O.H. Can the acute-phase reactant proteins be used as cancer biomarkers? Int. J. Biol. Markers 2010, 25, 1–11.
  83. Kang, S.-M.; Sung, H.-J.; Ahn, J.-M.; Park, J.-Y.; Lee, S.-Y.; Park, C.-S.; Cho, J.-Y. The Haptoglobin β chain as a supportive biomarker for human lung cancers. Mol. Biosyst. 2011, 7, 1167–1175.
  84. Sung, H.-J.; Ahn, J.-M.; Yoon, Y.-H.; Rhim, T.-Y.; Park, C.-S.; Park, J.-Y.; Lee, S.-Y.; Kim, J.-W.; Cho, J.-Y. Identification and Validation of SAA as a Potential Lung Cancer Biomarker and its Involvement in Metastatic Pathogenesis of Lung Cancer. J. Proteome Res. 2011, 10, 1383–1395.
  85. Jensen, L.; Whitehead, A.S. Regulation of serum amyloid A protein expression during the acute-phase response. Biochem. J. 1998, 334, 489–503.
  86. De Petris, L.; Orre, L.M.; Kanter, L.; Pernemalm, M.; Koyi, H.; Lewensohn, R.; Lehtiö, J. Tumor expression of S100A6 correlates with survival of patients with stage I non-small-cell lung cancer. Lung Cancer 2009, 63, 410–417.
  87. Pujol, J.L.; Grenier, J.; Daurès, J.P.; Daver, A.; Pujol, H.; Michel, F.B. Serum fragment of cytokeratin subunit 19 measured by CYFRA 21-1 immunoradiometric assay as a marker of lung cancer. Cancer Res. 1993, 53, 61–66.
  88. Bergqvist, M.; Brattström, D.; Hesselius, P.; Wiklund, B.; Silen, A.; Wagenius, G.; Brodin, O. Cytokeratin 8 and 18 fragments measured in serum and their relation to survival in patients with non-small cell lung cancer. Anticancer Res. 1999, 19, 1833–1836.
  89. Xu, B.J.; Gonzalez, A.L.; Kikuchi, T.; Yanagisawa, K.; Massion, P.P.; Wu, H.; Mason, S.E.; Olson, S.J.; Shyr, Y.; Carbone, D.P.; et al. MALDI-MS derived prognostic protein markers for resected non-small cell lung cancer. Proteom. Clin. Appl. 2008, 2, 1508–1517.
  90. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018, 359, 926–930.
  91. Zamay, T.N.; Zamay, G.S.; Kolovskaya, O.S.; Zukov, R.A.; Petrova, M.M.; Gargaun, A.; Berezovski, M.V.; Kichkailo, A.S. Current and Prospective Protein Biomarkers of Lung Cancer. Cancers 2017, 9, 155.
  92. Alcala, K.; Guida, F.; Johansson, M.; Johansson, M.; Robbins, H.A.; Smith-Byrne, K.; Stevens, V.; Zahed, H. Lung Cancer Cohort Consortium The Blood Proteome of Imminent Lung Cancer Diagnosis. medRxiv 2022.
  93. Mehan, M.R.; Williams, S.A.; Siegfried, J.M.; Bigbee, W.L.; Weissfeld, J.L.; Wilson, D.O.; Pass, H.I.; Rom, W.N.; Muley, T.; Meister, M.; et al. Validation of a blood protein signature for non-small cell lung cancer. Clin. Proteom. 2014, 11, 32.
  94. Boyle, P.; Chapman, C.J.; Holdenrieder, S.; Murray, A.; Robertson, C.; Wood, W.C.; Maddison, P.; Healey, G.; Fairley, G.H.; Barnes, A.C.; et al. Clinical validation of an autoantibody test for lung cancer. Ann. Oncol. 2011, 22, 383–389.
  95. Tan, E.M. Autoantibodies as reporters identifying aberrant cellular mechanisms in tumorigenesis. J. Clin. Investig. 2001, 108, 1411–1415.
  96. Houghton, A.N. Cancer antigens: Immune recognition of self and altered self. J. Exp. Med. 1994, 180, 1–4.
  97. Tan, E.M.; Zhang, J. Autoantibodies to tumor-associated antigens: Reporters from the immune system. Immunol. Rev. 2008, 222, 328–340.
  98. Zhang, J.-Y.; Tan, E.M. Autoantibodies to tumor-associated antigens as diagnostic biomarkers in hepatocellular carcinoma and other solid tumors. Expert Rev. Mol. Diagn. 2010, 10, 321–328.
  99. Du, Q.; Yu, R.; Wang, H.; Yan, N.; Yuan, Q.; Ma, Y.; Slamon, D.; Hou, D.; Wang, H.; Wang, Q. Significance of tumor-associated autoantibodies in the early diagnosis of lung cancer. Clin. Respir. J. 2018, 12, 2020–2028.
  100. Veronesi, G.; Bianchi, F.; Infante, M.; Alloisio, M. The challenge of small lung nodules identified in CT screening: Can biomarkers assist diagnosis? Biomark. Med. 2016, 10, 137–143.
  101. Chapman, C.; Murray, A.; Chakrabarti, J.; Thorpe, A.; Woolston, C.; Sahin, U.; Barnes, A.; Robertson, J. Autoantibodies in breast cancer: Their use as an aid to early diagnosis. Ann. Oncol. 2007, 18, 868–873.
  102. Dai, L.; Tsay, J.-C.J.; Li, J.; Yie, T.-A.; Munger, J.S.; Pass, H.; Rom, W.N.; Zhang, Y.; Tan, E.M.; Zhang, J.-Y. Autoantibodies against tumor-associated antigens in the early detection of lung cancer. Lung Cancer 2016, 99, 172–179.
  103. Burotto, M.; Thomas, A.; Subramaniam, D.; Giaccone, G.; Rajan, A. Biomarkers in Early-Stage Non–Small-Cell Lung Cancer: Current Concepts and Future Directions. J. Thorac. Oncol. 2014, 9, 1609–1617.
  104. Chen, H.; Werner, S.; Tao, S.; Zörnig, I.; Brenner, H. Blood autoantibodies against tumor-associated antigens as biomarkers in early detection of colorectal cancer. Cancer Lett. 2014, 346, 178–187.
  105. Lubin, R.; Zalcman, G.; Bouchet, L.; Trédaniel, J.; Legros, Y.; Cazals, D.; Hirsch, A.; Soussi, T. Serum p53 antibodies as early markers of lung cancer. Nat. Med. 1995, 1, 701–702.
  106. Murray, A.; Chapman, C.; Healey, G.; Peek, L.; Parsons, G.; Baldwin, D.; Barnes, A.; Sewell, H.; Fritsche, H.; Robertson, J. Technical validation of an autoantibody test for lung cancer. Ann. Oncol. 2010, 21, 1687–1693.
  107. Farlow, E.C.; Patel, K.; Basu, S.; Lee, B.S.; Kim, A.W.; Coon, J.S.; Faber, L.P.; Bonomi, P.; Liptay, M.J.; Borgia, J.A. Development of a Multiplexed Tumor-Associated Autoantibody-Based Blood Test for the Detection of Non–Small Cell Lung CancerEarly Detection Serum Test for NSCLC. Clin. Cancer Res. 2010, 16, 3452–3462.
  108. Wu, L.; Chang, W.; Zhao, J.; Yu, Y.; Tan, X.; Su, T.; Zhao, L.; Huang, S.; Liu, S.; Cao, G. Development of Autoantibody Signatures as Novel Diagnostic Biomarkers of Non–Small Cell Lung CancerNew Serum Diagnostic Biomarkers for NSCLC. Clin. Cancer Res. 2010, 16, 3760–3768.
  109. Lam, S.; Boyle, P.; Healey, G.F.; Maddison, P.; Peek, L.; Murray, A.; Chapman, C.J.; Allen, J.; Wood, W.C.; Sewell, H.F.; et al. EarlyCDT-Lung: An Immunobiomarker Test as an Aid to Early Detection of Lung Cancer. Cancer Prev. Res. 2011, 4, 1126–1134.
  110. Wang, T.; Liu, H.; Pei, L.; Wang, K.; Song, C.; Wang, P.; Ye, H.; Zhang, J.; Ji, Z.; Ouyang, S.; et al. Screening of tumor-associated antigens based on Oncomine database and evaluation of diagnostic value of autoantibodies in lung cancer. Clin. Immunol. 2020, 210, 108262.
More
This entry is offline, you can click here to edit this entry!
Video Production Service